101
|
Mapping Astrocyte Transcriptional Signatures in Response to Neuroactive Compounds. Int J Mol Sci 2021; 22:ijms22083975. [PMID: 33921461 PMCID: PMC8069033 DOI: 10.3390/ijms22083975] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 01/11/2023] Open
Abstract
Astrocytes play central roles in normal brain function and are critical components of synaptic networks that oversee behavioral outputs. Despite their close affiliation with neurons, how neuronal-derived signals influence astrocyte function at the gene expression level remains poorly characterized, largely due to difficulties associated with dissecting neuron- versus astrocyte-specific effects. Here, we use an in vitro system of stem cell-derived astrocytes to identify gene expression profiles in astrocytes that are influenced by neurons and regulate astrocyte development. Furthermore, we show that neurotransmitters and neuromodulators induce distinct transcriptomic and chromatin accessibility changes in astrocytes that are unique to each of these neuroactive compounds. These findings are highlighted by the observation that noradrenaline has a more profound effect on transcriptional profiles of astrocytes compared to glutamate, gamma-aminobutyric acid (GABA), acetylcholine, and serotonin. This is demonstrated through enhanced noradrenaline-induced transcriptomic and chromatin accessibility changes in vitro and through enhanced calcium signaling in vivo. Taken together, our study reveals distinct transcriptomic and chromatin architecture signatures in astrocytes in response to neuronal-derived neuroactive compounds. Since astrocyte function is affected in all neurological disorders, this study provides a new entry point for exploring genetic mechanisms of astrocyte-neuron communication that may be dysregulated in disease.
Collapse
|
102
|
Lim D, Semyanov A, Genazzani A, Verkhratsky A. Calcium signaling in neuroglia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:1-53. [PMID: 34253292 DOI: 10.1016/bs.ircmb.2021.01.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glial cells exploit calcium (Ca2+) signals to perceive the information about the activity of the nervous tissue and the tissue environment to translate this information into an array of homeostatic, signaling and defensive reactions. Astrocytes, the best studied glial cells, use several Ca2+ signaling generation pathways that include Ca2+ entry through plasma membrane, release from endoplasmic reticulum (ER) and from mitochondria. Activation of metabotropic receptors on the plasma membrane of glial cells is coupled to an enzymatic cascade in which a second messenger, InsP3 is generated thus activating intracellular Ca2+ release channels in the ER endomembrane. Astrocytes also possess store-operated Ca2+ entry and express several ligand-gated Ca2+ channels. In vivo astrocytes generate heterogeneous Ca2+ signals, which are short and frequent in distal processes, but large and relatively rare in soma. In response to neuronal activity intracellular and inter-cellular astrocytic Ca2+ waves can be produced. Astrocytic Ca2+ signals are involved in secretion, they regulate ion transport across cell membranes, and are contributing to cell morphological plasticity. Therefore, astrocytic Ca2+ signals are linked to fundamental functions of the central nervous system ranging from synaptic transmission to behavior. In oligodendrocytes, Ca2+ signals are generated by plasmalemmal Ca2+ influx, or by release from intracellular stores, or by combination of both. Microglial cells exploit Ca2+ permeable ionotropic purinergic receptors and transient receptor potential channels as well as ER Ca2+ release. In this contribution, basic morphology of glial cells, glial Ca2+ signaling toolkit, intracellular Ca2+ signals and Ca2+-regulated functions are discussed with focus on astrocytes.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Faculty of Biology, Moscow State University, Moscow, Russia; Sechenov First Moscow State Medical University, Moscow, Russia
| | - Armando Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Alexei Verkhratsky
- Sechenov First Moscow State Medical University, Moscow, Russia; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
103
|
Fischer T, Prey J, Eschholz L, Rotermund N, Lohr C. Norepinephrine-Induced Calcium Signaling and Store-Operated Calcium Entry in Olfactory Bulb Astrocytes. Front Cell Neurosci 2021; 15:639754. [PMID: 33833669 PMCID: PMC8021869 DOI: 10.3389/fncel.2021.639754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/02/2021] [Indexed: 11/30/2022] Open
Abstract
It is well-established that astrocytes respond to norepinephrine with cytosolic calcium rises in various brain areas, such as hippocampus or neocortex. However, less is known about the effect of norepinephrine on olfactory bulb astrocytes. In the present study, we used confocal calcium imaging and immunohistochemistry in mouse brain slices of the olfactory bulb, a brain region with a dense innervation of noradrenergic fibers, to investigate the calcium signaling evoked by norepinephrine in astrocytes. Our results show that application of norepinephrine leads to a cytosolic calcium rise in astrocytes which is independent of neuronal activity and mainly mediated by PLC/IP3-dependent internal calcium release. In addition, store-operated calcium entry (SOCE) contributes to the late phase of the response. Antagonists of both α1- and α2-adrenergic receptors, but not β-receptors, largely reduce the adrenergic calcium response, indicating that both α-receptor subtypes mediate norepinephrine-induced calcium transients in olfactory bulb astrocytes, whereas β-receptors do not contribute to the calcium transients.
Collapse
Affiliation(s)
- Timo Fischer
- Division of Neurophysiology, Department of Biology, Institute of Zoology, University of Hamburg, Hamburg, Germany
| | - Jessica Prey
- Division of Neurophysiology, Department of Biology, Institute of Zoology, University of Hamburg, Hamburg, Germany
| | - Lena Eschholz
- Division of Neurophysiology, Department of Biology, Institute of Zoology, University of Hamburg, Hamburg, Germany
| | - Natalie Rotermund
- Division of Neurophysiology, Department of Biology, Institute of Zoology, University of Hamburg, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, Department of Biology, Institute of Zoology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
104
|
Iwai Y, Ozawa K, Yahagi K, Mishima T, Akther S, Vo CT, Lee AB, Tanaka M, Itohara S, Hirase H. Transient Astrocytic Gq Signaling Underlies Remote Memory Enhancement. Front Neural Circuits 2021; 15:658343. [PMID: 33828463 PMCID: PMC8019746 DOI: 10.3389/fncir.2021.658343] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 01/31/2023] Open
Abstract
Astrocytes elicit transient Ca2+ elevations induced by G protein-coupled receptors (GPCRs), yet their role in vivo remains unknown. To address this, transgenic mice with astrocytic expression of the optogenetic Gq-type GPCR, Optoα1AR, were established, in which transient Ca2+ elevations similar to those in wild type mice were induced by brief blue light illumination. Activation of cortical astrocytes resulted in an adenosine A1 receptor-dependent inhibition of neuronal activity. Moreover, sensory stimulation with astrocytic activation induced long-term depression of sensory evoked response. At the behavioral level, repeated astrocytic activation in the anterior cortex gradually affected novel open field exploratory behavior, and remote memory was enhanced in a novel object recognition task. These effects were blocked by A1 receptor antagonism. Together, we demonstrate that GPCR-triggered Ca2+ elevation in cortical astrocytes has causal impacts on neuronal activity and behavior.
Collapse
Affiliation(s)
- Youichi Iwai
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
| | - Katsuya Ozawa
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
| | - Kazuko Yahagi
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
| | - Tsuneko Mishima
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sonam Akther
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Trang Vo
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ashley Bomin Lee
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mika Tanaka
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Wako, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Wako, Japan
| | - Hajime Hirase
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
105
|
Vaidyanathan TV, Collard M, Yokoyama S, Reitman ME, Poskanzer KE. Cortical astrocytes independently regulate sleep depth and duration via separate GPCR pathways. eLife 2021; 10:63329. [PMID: 33729913 PMCID: PMC7968927 DOI: 10.7554/elife.63329] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Non-rapid eye movement (NREM) sleep, characterized by slow-wave electrophysiological activity, underlies several critical functions, including learning and memory. However, NREM sleep is heterogeneous, varying in duration, depth, and spatially across the cortex. While these NREM sleep features are thought to be largely independently regulated, there is also evidence that they are mechanistically coupled. To investigate how cortical NREM sleep features are controlled, we examined the astrocytic network, comprising a cortex-wide syncytium that influences population-level neuronal activity. We quantified endogenous astrocyte activity in mice over natural sleep and wake, then manipulated specific astrocytic G-protein-coupled receptor (GPCR) signaling pathways in vivo. We find that astrocytic Gi- and Gq-coupled GPCR signaling separately control NREM sleep depth and duration, respectively, and that astrocytic signaling causes differential changes in local and remote cortex. These data support a model in which the cortical astrocyte network serves as a hub for regulating distinct NREM sleep features. Sleep has many roles, from strengthening new memories to regulating mood and appetite. While we might instinctively think of sleep as a uniform state of reduced brain activity, the reality is more complex. First, over the course of the night, we cycle between a number of different sleep stages, which reflect different levels of sleep depth. Second, the amount of sleep depth is not necessarily even across the brain but can vary between regions. These sleep stages consist of either rapid eye movement (REM) sleep or non-REM (NREM) sleep. REM sleep is when most dreaming occurs, whereas NREM sleep is particularly important for learning and memory and can vary in duration and depth. During NREM sleep, large groups of neurons synchronize their firing to create rhythmic waves of activity known as slow waves. The more synchronous the activity, the deeper the sleep. Vaidyanathan et al. now show that brain cells called astrocytes help regulate NREM sleep. Astrocytes are not neurons but belong to a group of specialized cells called glia. They are the largest glia cell type in the brain and display an array of proteins on their surfaces called G-protein-coupled receptors (GPCRs). These enable them to sense sleep-wake signals from other parts of the brain and to generate their own signals. In fact, each astrocyte can communicate with thousands of neurons at once. They are therefore well-poised to coordinate brain activity during NREM sleep. Using innovative tools, Vaidyanathan et al. visualized astrocyte activity in mice as the animals woke up or fell asleep. The results showed that astrocytes change their activity just before each sleep–wake transition. They also revealed that astrocytes control both the depth and duration of NREM sleep via two different types of GPCR signals. Increasing one of these signals (Gi-GPCR) made the mice sleep more deeply but did not change sleep duration. Decreasing the other (Gq-GPCR) made the mice sleep for longer but did not affect sleep depth. Sleep problems affect many people at some point in their lives, and often co-exist with other conditions such as mental health disorders. Understanding how the brain regulates different features of sleep could help us develop better – and perhaps more specific – treatments for sleep disorders. The current study suggests that manipulating GPCRs on astrocytes might increase sleep depth, for example. But before work to test this idea can begin, we must first determine whether findings from sleeping mice also apply to people.
Collapse
Affiliation(s)
- Trisha V Vaidyanathan
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Max Collard
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Sae Yokoyama
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Michael E Reitman
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States
| | - Kira E Poskanzer
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, San Francisco, United States
| |
Collapse
|
106
|
Bellot-Saez A, Stevenson R, Kékesi O, Samokhina E, Ben-Abu Y, Morley JW, Buskila Y. Neuromodulation of Astrocytic K + Clearance. Int J Mol Sci 2021; 22:ijms22052520. [PMID: 33802343 PMCID: PMC7959145 DOI: 10.3390/ijms22052520] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/21/2022] Open
Abstract
Potassium homeostasis is fundamental for brain function. Therefore, effective removal of excessive K+ from the synaptic cleft during neuronal activity is paramount. Astrocytes play a key role in K+ clearance from the extracellular milieu using various mechanisms, including uptake via Kir channels and the Na+-K+ ATPase, and spatial buffering through the astrocytic gap-junction coupled network. Recently we showed that alterations in the concentrations of extracellular potassium ([K+]o) or impairments of the astrocytic clearance mechanism affect the resonance and oscillatory behavior of both the individual and networks of neurons. These results indicate that astrocytes have the potential to modulate neuronal network activity, however, the cellular effectors that may affect the astrocytic K+ clearance process are still unknown. In this study, we have investigated the impact of neuromodulators, which are known to mediate changes in network oscillatory behavior, on the astrocytic clearance process. Our results suggest that while some neuromodulators (5-HT; NA) might affect astrocytic spatial buffering via gap-junctions, others (DA; Histamine) primarily affect the uptake mechanism via Kir channels. These results suggest that neuromodulators can affect network oscillatory activity through parallel activation of both neurons and astrocytes, establishing a synergistic mechanism to maximize the synchronous network activity.
Collapse
Affiliation(s)
- Alba Bellot-Saez
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Rebecca Stevenson
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Orsolya Kékesi
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Evgeniia Samokhina
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Yuval Ben-Abu
- Projects and Physics Section, Sapir Academic College, D.N. Hof Ashkelon, Sderot 79165, Israel;
| | - John W. Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
- International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW 2751, Australia
- Correspondence: ; Tel.: +61-246203853
| |
Collapse
|
107
|
Wahis J, Holt MG. Astrocytes, Noradrenaline, α1-Adrenoreceptors, and Neuromodulation: Evidence and Unanswered Questions. Front Cell Neurosci 2021; 15:645691. [PMID: 33716677 PMCID: PMC7947346 DOI: 10.3389/fncel.2021.645691] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/03/2021] [Indexed: 12/27/2022] Open
Abstract
Noradrenaline is a major neuromodulator in the central nervous system (CNS). It is released from varicosities on neuronal efferents, which originate principally from the main noradrenergic nuclei of the brain - the locus coeruleus - and spread throughout the parenchyma. Noradrenaline is released in response to various stimuli and has complex physiological effects, in large part due to the wide diversity of noradrenergic receptors expressed in the brain, which trigger diverse signaling pathways. In general, however, its main effect on CNS function appears to be to increase arousal state. Although the effects of noradrenaline have been researched extensively, the majority of studies have assumed that noradrenaline exerts its effects by acting directly on neurons. However, neurons are not the only cells in the CNS expressing noradrenaline receptors. Astrocytes are responsive to a range of neuromodulators - including noradrenaline. In fact, noradrenaline evokes robust calcium transients in astrocytes across brain regions, through activation of α1-adrenoreceptors. Crucially, astrocytes ensheath neurons at synapses and are known to modulate synaptic activity. Hence, astrocytes are in a key position to relay, or amplify, the effects of noradrenaline on neurons, most notably by modulating inhibitory transmission. Based on a critical appraisal of the current literature, we use this review to argue that a better understanding of astrocyte-mediated noradrenaline signaling is therefore essential, if we are ever to fully understand CNS function. We discuss the emerging concept of astrocyte heterogeneity and speculate on how this might impact the noradrenergic modulation of neuronal circuits. Finally, we outline possible experimental strategies to clearly delineate the role(s) of astrocytes in noradrenergic signaling, and neuromodulation in general, highlighting the urgent need for more specific and flexible experimental tools.
Collapse
Affiliation(s)
- Jérôme Wahis
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
108
|
Chemogenetic manipulation of astrocytic activity: Is it possible to reveal the roles of astrocytes? Biochem Pharmacol 2021; 186:114457. [PMID: 33556341 DOI: 10.1016/j.bcp.2021.114457] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 01/08/2023]
Abstract
Astrocytes are the major glial cells in the central nervous system, but unlike neurons, they do not produce action potentials. For many years, astrocytes were considered supporting cells in the central nervous system (CNS). Technological advances over the last two decades are changing the face of glial research. Accumulating data from recent investigations show that astrocytes display transient calcium spikes and regulate synaptic transmission by releasing transmitters called gliotransmitters. Many new powerful technologies are used to interfere with astrocytic activity, in order to obtain a better understanding of the roles of astrocytes in the brain. Among these technologies, chemogenetics has recently been used frequently. In this review, we will summarize new functions of astrocytes in the brain that have been revealed using this cutting-edge technique. Moreover, we will discuss the possibilities and challenges of manipulating astrocytic activity using this technology.
Collapse
|
109
|
Cortical astrocytes regulate ethanol consumption and intoxication in mice. Neuropsychopharmacology 2021; 46:500-508. [PMID: 32464636 PMCID: PMC8027025 DOI: 10.1038/s41386-020-0721-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are fundamental building blocks of the central nervous system. Their dysfunction has been implicated in many psychiatric disorders, including alcohol use disorder, yet our understanding of their functional role in ethanol intoxication and consumption is very limited. Astrocytes regulate behavior through multiple intracellular signaling pathways, including G-protein coupled-receptor (GPCR)-mediated calcium signals. To test the hypothesis that GPCR-induced calcium signaling is also involved in the behavioral effects of ethanol, we expressed astrocyte-specific excitatory DREADDs in the prefrontal cortex (PFC) of mice. Activating Gq-GPCR signaling in PFC astrocytes increased drinking in ethanol-naïve mice, but not in mice with a history of ethanol drinking. In contrast, reducing calcium signaling with an astrocyte-specific calcium extruder reduced ethanol intake. Cortical astrocyte calcium signaling also altered the acute stimulatory and sedative-hypnotic effects of ethanol. Astrocyte-specific Gq-DREADD activation increased both the locomotor-activating effects of low dose ethanol and the sedative-hypnotic effects of a high dose, while reduced astrocyte calcium signaling diminished sensitivity to the hypnotic effects. In addition, we found that adenosine A1 receptors were required for astrocyte calcium activation to increase ethanol sedation. These results support integral roles for PFC astrocytes in the behavioral actions of ethanol that are due, at least in part, to adenosine receptor activation.
Collapse
|
110
|
Aboufares El Alaoui A, Jackson M, Fabri M, de Vivo L, Bellesi M. Characterization of Subcellular Organelles in Cortical Perisynaptic Astrocytes. Front Cell Neurosci 2021; 14:573944. [PMID: 33633542 PMCID: PMC7901967 DOI: 10.3389/fncel.2020.573944] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/30/2020] [Indexed: 01/18/2023] Open
Abstract
Perisynaptic astrocytic processes (PAPs) carry out several different functions, from metabolite clearing to control of neuronal excitability and synaptic plasticity. All these functions are likely orchestrated by complex cellular machinery that resides within the PAPs and relies on a fine interplay between multiple subcellular components. However, traditional transmission electron microscopy (EM) studies have found that PAPs are remarkably poor of intracellular organelles, failing to explain how such a variety of PAP functions are achieved in the absence of a proportional complex network of intracellular structures. Here, we use serial block-face scanning EM to reconstruct and describe in three dimensions PAPs and their intracellular organelles in two different mouse cortical regions. We described five distinct organelles, which included empty and full endosomes, phagosomes, mitochondria, and endoplasmic reticulum (ER) cisternae, distributed within three PAPs categories (branches, branchlets, and leaflets). The majority of PAPs belonged to the leaflets category (~60%), with branchlets representing a minority (~37%). Branches were rarely in contact with synapses (<3%). Branches had a higher density of mitochondria and ER cisternae than branchlets and leaflets. Also, branches and branchlets displayed organelles more frequently than leaflets. Endosomes and phagosomes, which accounted for more than 60% of all the organelles detected, were often associated with the same PAP. Likewise, mitochondria and ER cisternae, representing ~40% of all organelles were usually associated. No differences were noted between the organelle distribution of the somatosensory and the anterior cingulate cortex. Finally, the organelle distribution in PAPs did not largely depend on the presence of a spine apparatus or a pre-synaptic mitochondrion in the synapse that PAPs were enwrapping, with some exceptions regarding the presence of phagosomes and ER cisternae, which were slightly more represented around synapses lacking a spine apparatus and a presynaptic mitochondrion, respectively. Thus, PAPs contain several subcellular organelles that could underlie the diverse astrocytic functions carried out at central synapses.
Collapse
Affiliation(s)
- Amina Aboufares El Alaoui
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy.,School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Molly Jackson
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Mara Fabri
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Luisa de Vivo
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Michele Bellesi
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
111
|
Nagai J, Yu X, Papouin T, Cheong E, Freeman MR, Monk KR, Hastings MH, Haydon PG, Rowitch D, Shaham S, Khakh BS. Behaviorally consequential astrocytic regulation of neural circuits. Neuron 2020; 109:576-596. [PMID: 33385325 DOI: 10.1016/j.neuron.2020.12.008] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Astrocytes are a large and diverse population of morphologically complex cells that exist throughout nervous systems of multiple species. Progress over the last two decades has shown that astrocytes mediate developmental, physiological, and pathological processes. However, a long-standing open question is how astrocytes regulate neural circuits in ways that are behaviorally consequential. In this regard, we summarize recent studies using Caenorhabditis elegans, Drosophila melanogaster, Danio rerio, and Mus musculus. The data reveal diverse astrocyte mechanisms operating in seconds or much longer timescales within neural circuits and shaping multiple behavioral outputs. We also refer to human diseases that have a known primary astrocytic basis. We suggest that including astrocytes in mechanistic, theoretical, and computational studies of neural circuits provides new perspectives to understand behavior, its regulation, and its disease-related manifestations.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; RIKEN Center for Brain Science, 2-1 Hirosawa Wako City, Saitama 351-0198, Japan
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 514 Burrill Hall, 407 S. Goodwin Ave, Urbana, IL 61801, USA
| | - Thomas Papouin
- Department of Neuroscience, Washington University in St. Louis, School of Medicine, Campus Box 8108, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Marc R Freeman
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Kelly R Monk
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - David Rowitch
- Department of Paediatrics, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Departments of Pediatrics and Neurosurgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
112
|
Sharma K, Gordon GRJ, Tran CHT. Heterogeneity of Sensory-Induced Astrocytic Ca 2+ Dynamics During Functional Hyperemia. Front Physiol 2020; 11:611884. [PMID: 33362585 PMCID: PMC7758506 DOI: 10.3389/fphys.2020.611884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022] Open
Abstract
Astrocytic Ca2+ fluctuations associated with functional hyperemia have typically been measured from large cellular compartments such as the soma, the whole arbor and the endfoot. The most prominent Ca2+ event is a large magnitude, delayed signal that follows vasodilation. However, previous work has provided little information about the spatio-temporal properties of such Ca2+ transients or their heterogeneity. Here, using an awake, in vivo two-photon fluorescence-imaging model, we performed detailed profiling of delayed astrocytic Ca2+ signals across astrocytes or within individual astrocyte compartments using small regions of interest next to penetrating arterioles and capillaries along with vasomotor responses to vibrissae stimulation. We demonstrated that while a 5-s air puff that stimulates all whiskers predominantly generated reproducible functional hyperemia in the presence or absence of astrocytic Ca2+ changes, whisker stimulation inconsistently produced astrocytic Ca2+ responses. More importantly, these Ca2+ responses were heterogeneous among subcellular structures of the astrocyte and across different astrocytes that resided within the same field of view. Furthermore, we found that whisker stimulation induced discrete Ca2+ “hot spots” that spread regionally within the endfoot. These data reveal that astrocytic Ca2+ dynamics associated with the microvasculature are more complex than previously thought, and highlight the importance of considering the heterogeneity of astrocytic Ca2+ activity to fully understanding neurovascular coupling.
Collapse
Affiliation(s)
- Kushal Sharma
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - Grant R J Gordon
- Department of Physiology and Pharmacology, School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Cam Ha T Tran
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV, United States
| |
Collapse
|
113
|
Qin H, He W, Yang C, Li J, Jian T, Liang S, Chen T, Feng H, Chen X, Liao X, Zhang K. Monitoring Astrocytic Ca 2+ Activity in Freely Behaving Mice. Front Cell Neurosci 2020; 14:603095. [PMID: 33343304 PMCID: PMC7744696 DOI: 10.3389/fncel.2020.603095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
Monitoring astrocytic Ca2+ activity is essential to understand the physiological and pathological roles of astrocytes in the brain. However, previous commonly used methods for studying astrocytic Ca2+ activities can be applied in only anesthetized or head-fixed animals, which significantly affects in vivo astrocytic Ca2+ dynamics. In the current study, we combined optic fiber recordings with genetically encoded Ca2+ indicators (GECIs) to monitor astrocytic activity in freely behaving mice. This approach enabled selective and reliable measurement of astrocytic Ca2+ activity, which was verified by the astrocyte-specific labeling of GECIs and few movement artifacts. Additionally, astrocytic Ca2+ activities induced by locomotion or footshock were stably recorded in the cortices and hippocampi of freely behaving mice. Furthermore, this method allowed for the longitudinal study of astrocytic activities over several weeks. This work provides a powerful approach to record astrocytic activity selectively, stably, and chronically in freely behaving mice.
Collapse
Affiliation(s)
- Han Qin
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China.,Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Wenjing He
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Chuanyan Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jin Li
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Tingliang Jian
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Shanshan Liang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Tunan Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
114
|
Ryczko D, Hanini‐Daoud M, Condamine S, Bréant BJB, Fougère M, Araya R, Kolta A. S100β‐mediated astroglial control of firing and input processing in layer 5 pyramidal neurons of the mouse visual cortex. J Physiol 2020; 599:677-707. [DOI: 10.1113/jp280501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Dimitri Ryczko
- Département de Neurosciences Université de Montréal Montréal QC Canada
- Département de Pharmacologie‐Physiologie Université de Sherbrooke Sherbrooke QC Canada
- Centre de recherche du CHUS Sherbrooke QC Canada
- Institut de Pharmacologie de Sherbrooke Sherbrooke QC Canada
- Centre d'excellence en neurosciences de l'Université de Sherbrooke Sherbrooke QC Canada
| | | | - Steven Condamine
- Département de Neurosciences Université de Montréal Montréal QC Canada
| | | | - Maxime Fougère
- Département de Pharmacologie‐Physiologie Université de Sherbrooke Sherbrooke QC Canada
| | - Roberto Araya
- Département de Neurosciences Université de Montréal Montréal QC Canada
| | - Arlette Kolta
- Département de Neurosciences Université de Montréal Montréal QC Canada
- Faculté de Médecine Dentaire Université de Montréal Montréal QC Canada
| |
Collapse
|
115
|
Shen W, Chen S, Xiang Y, Yao Z, Chen Z, Wu X, Li L, Zeng LH. Astroglial adrenoreceptors modulate synaptic transmission and contextual fear memory formation in dentate gyrus. Neurochem Int 2020; 143:104942. [PMID: 33340594 DOI: 10.1016/j.neuint.2020.104942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/30/2020] [Accepted: 12/13/2020] [Indexed: 11/16/2022]
Abstract
Astrocytes perform various supporting functions, including ion buffering, metabolic supplying and neurotransmitter clearance. They can also sense neuronal activity owing to the presence of specific receptors for neurotransmitters. In turn, astrocytes can regulate synaptic activity through the release of gliotransmitters. Evidence has shown that astrocytes are very sensitive to the locus coeruleus (LC) afferents. However, little is known about how LC neuromodulatory norepinephrine (NE) modulates synaptic transmission through astrocytic activity. In mouse dentate gyrus (DG), we demonstrated an increase in the frequency of miniature excitatory postsynaptic currents (mEPSC) in response to NE, which required the release of glutamate from astrocytes. The rise in glutamate release probability is likely due to the activation of presynaptic GluN2B-containing NMDA receptors. Moreover, we showed that the activation of NE signaling in DG is necessary for the formation of contextual learning memory. Thus, NE signaling activation during fear conditioning training contributed to enduring changes in the frequency of mEPSC in DG. Our results strongly support the physiological neuromodulatory role of NE signaling, which is derived from activation of astrocytes.
Collapse
Affiliation(s)
- Weida Shen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China.
| | - Shishuo Chen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China; Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yingchun Xiang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zheyu Yao
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Zhitao Chen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Xitian Wu
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Ling Li
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Ling-Hui Zeng
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China; Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
116
|
Ma Z, Freeman MR. TrpML-mediated astrocyte microdomain Ca 2+ transients regulate astrocyte-tracheal interactions. eLife 2020; 9:e58952. [PMID: 33284108 PMCID: PMC7721441 DOI: 10.7554/elife.58952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/11/2020] [Indexed: 01/06/2023] Open
Abstract
Astrocytes exhibit spatially-restricted near-membrane microdomain Ca2+transients in their fine processes. How these transients are generated and regulate brain function in vivo remains unclear. Here we show that Drosophila astrocytes exhibit spontaneous, activity-independent microdomain Ca2+ transients in their fine processes. Astrocyte microdomain Ca2+ transients are mediated by the TRP channel TrpML, stimulated by reactive oxygen species (ROS), and can be enhanced in frequency by the neurotransmitter tyramine via the TyrRII receptor. Interestingly, many astrocyte microdomain Ca2+ transients are closely associated with tracheal elements, which dynamically extend filopodia throughout the central nervous system (CNS) to deliver O2 and regulate gas exchange. Many astrocyte microdomain Ca2+ transients are spatio-temporally correlated with the initiation of tracheal filopodial retraction. Loss of TrpML leads to increased tracheal filopodial numbers, growth, and increased CNS ROS. We propose that local ROS production can activate astrocyte microdomain Ca2+ transients through TrpML, and that a subset of these microdomain transients promotes tracheal filopodial retraction and in turn modulate CNS gas exchange.
Collapse
Affiliation(s)
- Zhiguo Ma
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Marc R Freeman
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
117
|
Ye L, Orynbayev M, Zhu X, Lim EY, Dereddi RR, Agarwal A, Bergles DE, Bhat MA, Paukert M. Ethanol abolishes vigilance-dependent astroglia network activation in mice by inhibiting norepinephrine release. Nat Commun 2020; 11:6157. [PMID: 33268792 PMCID: PMC7710743 DOI: 10.1038/s41467-020-19475-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Norepinephrine adjusts sensory processing in cortical networks and gates plasticity enabling adaptive behavior. The actions of norepinephrine are profoundly altered by recreational drugs like ethanol, but the consequences of these changes on distinct targets such as astrocytes, which exhibit norepinephrine-dependent Ca2+ elevations during vigilance, are not well understood. Using in vivo two-photon imaging, we show that locomotion-induced Ca2+ elevations in mouse astroglia are profoundly inhibited by ethanol, an effect that can be reversed by enhancing norepinephrine release. Vigilance-dependent astroglial activation is abolished by deletion of α1A-adrenergic receptor from astroglia, indicating that norepinephrine acts directly on these ubiquitous glial cells. Ethanol reduces vigilance-dependent Ca2+ transients in noradrenergic terminals, but has little effect on astroglial responsiveness to norepinephrine, suggesting that ethanol suppresses their activation by inhibiting norepinephrine release. Since abolition of astroglia Ca2+ activation does not affect motor coordination, global suppression of astroglial networks may contribute to the cognitive effects of alcohol intoxication. The effects of norepinephrine on sensory processing in cortical networks are altered by recreational drugs like ethanol. The authors show that ethanol suppresses the activation of astrocytes by inhibiting norepinephrine release which may contribute to the cognitive effects of alcohol intoxication.
Collapse
Affiliation(s)
- Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Murat Orynbayev
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiangyu Zhu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Eunice Y Lim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ram R Dereddi
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
118
|
Ingiosi AM, Hayworth CR, Harvey DO, Singletary KG, Rempe MJ, Wisor JP, Frank MG. A Role for Astroglial Calcium in Mammalian Sleep and Sleep Regulation. Curr Biol 2020; 30:4373-4383.e7. [PMID: 32976809 PMCID: PMC7919541 DOI: 10.1016/j.cub.2020.08.052] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 07/07/2020] [Accepted: 08/13/2020] [Indexed: 10/23/2022]
Abstract
Mammalian sleep expression and regulation have historically been thought to reflect the activity of neurons. Changes in other brain cells (glia) across the sleep-wake cycle and their role in sleep regulation are comparatively unexplored. We show that sleep and wakefulness are accompanied by state-dependent changes in astroglial activity. Using a miniature microscope in freely behaving mice and a two-photon microscope in head-fixed, unanesthetized mice, we show that astroglial calcium signals are highest in wake and lowest in sleep and are most pronounced in astroglial processes. We also find that astroglial calcium signals during non-rapid eye movement sleep change in proportion to sleep need. In contrast to neurons, astrocytes become less synchronized during non-rapid eye movement sleep after sleep deprivation at the network and single-cell level. Finally, we show that conditionally reducing intracellular calcium in astrocytes impairs the homeostatic response to sleep deprivation. Thus, astroglial calcium activity changes dynamically across vigilance states, is proportional to sleep need, and is a component of the sleep homeostat.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA
| | - Christopher R Hayworth
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA
| | - Daniel O Harvey
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA
| | - Kristan G Singletary
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA
| | - Michael J Rempe
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA; Department of Mathematics and Computer Science, Whitworth University, West Hawthorne Road, Spokane, WA 99251, USA
| | - Jonathan P Wisor
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA
| | - Marcos G Frank
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, East Spokane Falls Boulevard, Spokane, WA 99202, USA.
| |
Collapse
|
119
|
Laing C, Blanchard N, McConkey GA. Noradrenergic Signaling and Neuroinflammation Crosstalk Regulate Toxoplasma gondii-Induced Behavioral Changes. Trends Immunol 2020; 41:1072-1082. [PMID: 33214056 DOI: 10.1016/j.it.2020.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022]
Abstract
Infections of the nervous system elicit neuroimmune responses and alter neurotransmission, affecting host neurological functions. Chronic infection with the apicomplexan parasite Toxoplasma correlates with certain neurological disorders in humans and alters behavior in rodents. Here, we propose that the crosstalk between neurotransmission and neuroinflammation may underlie some of these cognitive changes. We discuss how T. gondii infection suppresses noradrenergic signaling and how the restoration of this pathway improves behavioral aberrations, suggesting that altered neurotransmission and neuroimmune responses may act in concert to perturb behavior. This interaction might apply to other infectious agents, such as viruses, that elicit cognitive changes. We hypothesize that neurotransmitter signaling in immune cells can contribute to behavioral changes associated with brain infection, offering opportunities for potential therapeutic targeting.
Collapse
Affiliation(s)
- Conor Laing
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Nicolas Blanchard
- Centre de Physiopathologie Toulouse Purpan (CPTP), Inserm, CNRS, Université de Toulouse, Toulouse, France.
| | - Glenn A McConkey
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
120
|
Augusto-Oliveira M, Arrifano GP, Takeda PY, Lopes-Araújo A, Santos-Sacramento L, Anthony DC, Verkhratsky A, Crespo-Lopez ME. Astroglia-specific contributions to the regulation of synapses, cognition and behaviour. Neurosci Biobehav Rev 2020; 118:331-357. [DOI: 10.1016/j.neubiorev.2020.07.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
|
121
|
Williams CA, Miller KE, Williams NP, Portfors CV, Perkel DJ. Distribution and co-expression of adrenergic receptor-encoding mRNA in the mouse inferior colliculus. J Comp Neurol 2020; 529:1743-1755. [PMID: 33067825 DOI: 10.1002/cne.25053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 11/06/2022]
Abstract
Adrenergic receptors are mediators of adrenergic and noradrenergic modulation throughout the brain. Previous studies have provided evidence for the expression of adrenergic receptors in the midbrain auditory nucleus, the inferior colliculus (IC), but have not examined the cellular patterns of expression in detail. Here, we utilize multichannel fluorescent in situ hybridization to detect the expression of adrenergic receptor-encoding mRNA in the inferior colliculus of male and female mice. We found expression of α1 , α2A , and β2 receptor-encoding mRNA throughout all areas of the IC. While we observed similar levels of expression of α1 receptor-encoding mRNA across the subregions of the IC, α2A and β2 receptor-encoding mRNA was expressed differentially. To account for developmental changes in noradrenergic receptor expression, we measured expression levels in mice aged P15, P20, and P60. We observed little change in levels of expression across these ages. To ascertain the modulatory potential of multiple adrenergic receptor subtypes in a single IC cell, we measured co-expression of α1 , α2A , and β2 receptor-encoding mRNA. We found greater proportions of cells in the IC that expressed no adrenergic receptor-encoding mRNA, α1 and α2A adrenergic receptor-encoding mRNA, and α1, α2A, and β2 receptor-encoding mRNA than would be predicted by independent expression of each receptor subtype. These data suggest a coordinated pattern of adrenergic receptor expression in the IC and provide the first evidence for adrenergic receptor expression and co-expression in the subregions of the mouse auditory midbrain.
Collapse
Affiliation(s)
- Charles A Williams
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Kimberly E Miller
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Nisa P Williams
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Christine V Portfors
- Integrative Physiology and Neuroscience & School of Biological Sciences, Washington State University Vancouver, Vancouver, Washington, USA
| | - David J Perkel
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Otolaryngology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
122
|
Trujillo-Estrada L, Gomez-Arboledas A, Forner S, Martini AC, Gutierrez A, Baglietto-Vargas D, LaFerla FM. Astrocytes: From the Physiology to the Disease. Curr Alzheimer Res 2020; 16:675-698. [PMID: 31470787 DOI: 10.2174/1567205016666190830110152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
Abstract
Astrocytes are key cells for adequate brain formation and regulation of cerebral blood flow as well as for the maintenance of neuronal metabolism, neurotransmitter synthesis and exocytosis, and synaptic transmission. Many of these functions are intrinsically related to neurodegeneration, allowing refocusing on the role of astrocytes in physiological and neurodegenerative states. Indeed, emerging evidence in the field indicates that abnormalities in the astrocytic function are involved in the pathogenesis of multiple neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD) and Amyotrophic Lateral Sclerosis (ALS). In the present review, we highlight the physiological role of astrocytes in the CNS, including their communication with other cells in the brain. Furthermore, we discuss exciting findings and novel experimental approaches that elucidate the role of astrocytes in multiple neurological disorders.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Stefânia Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Antonia Gutierrez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| |
Collapse
|
123
|
Rasmussen R, O'Donnell J, Ding F, Nedergaard M. Interstitial ions: A key regulator of state-dependent neural activity? Prog Neurobiol 2020; 193:101802. [PMID: 32413398 PMCID: PMC7331944 DOI: 10.1016/j.pneurobio.2020.101802] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 02/08/2023]
Abstract
Throughout the nervous system, ion gradients drive fundamental processes. Yet, the roles of interstitial ions in brain functioning is largely forgotten. Emerging literature is now revitalizing this area of neuroscience by showing that interstitial cations (K+, Ca2+ and Mg2+) are not static quantities but change dynamically across states such as sleep and locomotion. In turn, these state-dependent changes are capable of sculpting neuronal activity; for example, changing the local interstitial ion composition in the cortex is sufficient for modulating the prevalence of slow-frequency neuronal oscillations, or potentiating the gain of visually evoked responses. Disturbances in interstitial ionic homeostasis may also play a central role in the pathogenesis of central nervous system diseases. For example, impairments in K+ buffering occur in a number of neurodegenerative diseases, and abnormalities in neuronal activity in disease models disappear when interstitial K+ is normalized. Here we provide an overview of the roles of interstitial ions in physiology and pathology. We propose the brain uses interstitial ion signaling as a global mechanism to coordinate its complex activity patterns, and ion homeostasis failure contributes to central nervous system diseases affecting cognitive functions and behavior.
Collapse
Affiliation(s)
- Rune Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - John O'Donnell
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - Fengfei Ding
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, United States.
| |
Collapse
|
124
|
Okubo Y. Astrocytic Ca2+ signaling mediated by the endoplasmic reticulum in health and disease. J Pharmacol Sci 2020; 144:83-88. [DOI: 10.1016/j.jphs.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
|
125
|
Pacholko AG, Wotton CA, Bekar LK. Astrocytes-The Ultimate Effectors of Long-Range Neuromodulatory Networks? Front Cell Neurosci 2020; 14:581075. [PMID: 33192327 PMCID: PMC7554522 DOI: 10.3389/fncel.2020.581075] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022] Open
Abstract
It was long thought that astrocytes, given their lack of electrical signaling, were not involved in communication with neurons. However, we now know that one astrocyte on average maintains and regulates the extracellular neurotransmitter and potassium levels of more than 140,000 synapses, both excitatory and inhibitory, within their individual domains, and form a syncytium that can propagate calcium waves to affect distant cells via release of “gliotransmitters” such as glutamate, ATP, or adenosine. Neuromodulators can affect signal-to-noise and frequency transmission within cortical circuits by effects on inhibition, allowing for the filtering of relevant vs. irrelevant stimuli. Moreover, synchronized “resting” and desynchronized “activated” brain states are gated by short bursts of high-frequency neuromodulatory activity, highlighting the need for neuromodulation that is robust, rapid, and far-reaching. As many neuromodulators are released in a volume manner where degradation/uptake and the confines of the complex CNS limit diffusion distance, we ask the question—are astrocytes responsible for rapidly extending neuromodulator actions to every synapse? Neuromodulators are known to influence transitions between brain states, leading to control over plasticity, responses to salient stimuli, wakefulness, and sleep. These rapid and wide-spread state transitions demand that neuromodulators can simultaneously influence large and diverse regions in a manner that should be impossible given the limitations of simple diffusion. Intriguingly, astrocytes are ideally situated to amplify/extend neuromodulator effects over large populations of synapses given that each astrocyte can: (1) ensheath a large number of synapses; (2) release gliotransmitters (glutamate/ATP/adenosine) known to affect inhibition; (3) regulate extracellular potassium that can affect excitability and excitation/inhibition balance; and (4) express receptors for all neuromodulators. In this review article, we explore the hypothesis that astrocytes extend and amplify neuromodulatory influences on neuronal networks via alterations in calcium dynamics, the release of gliotransmitters, and potassium homeostasis. Given that neuromodulatory networks are at the core of our sleep-wake cycle and behavioral states, and determine how we interact with our environment, this review article highlights the importance of basic astrocyte function in homeostasis, general cognition, and psychiatric disorders.
Collapse
Affiliation(s)
- Anthony G Pacholko
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Caitlin A Wotton
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lane K Bekar
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
126
|
Perez DM. α 1-Adrenergic Receptors in Neurotransmission, Synaptic Plasticity, and Cognition. Front Pharmacol 2020; 11:581098. [PMID: 33117176 PMCID: PMC7553051 DOI: 10.3389/fphar.2020.581098] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
α1-adrenergic receptors are G-Protein Coupled Receptors that are involved in neurotransmission and regulate the sympathetic nervous system through binding and activating the neurotransmitter, norepinephrine, and the neurohormone, epinephrine. There are three α1-adrenergic receptor subtypes (α1A, α1B, α1D) that are known to play various roles in neurotransmission and cognition. They are related to two other adrenergic receptor families that also bind norepinephrine and epinephrine, the β- and α2-, each with three subtypes (β1, β2, β3, α2A, α2B, α2C). Previous studies assessing the roles of α1-adrenergic receptors in neurotransmission and cognition have been inconsistent. This was due to the use of poorly-selective ligands and many of these studies were published before the characterization of the cloned receptor subtypes and the subsequent development of animal models. With the availability of more-selective ligands and the development of animal models, a clearer picture of their role in cognition and neurotransmission can be assessed. In this review, we highlight the significant role that the α1-adrenergic receptor plays in regulating synaptic efficacy, both short and long-term synaptic plasticity, and its regulation of different types of memory. We will also present evidence that the α1-adrenergic receptors, and particularly the α1A-adrenergic receptor subtype, are a potentially good target to treat a wide variety of neurological conditions with diminished cognition.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
127
|
Kwak H, Koh W, Kim S, Song K, Shin JI, Lee JM, Lee EH, Bae JY, Ha GE, Oh JE, Park YM, Kim S, Feng J, Lee SE, Choi JW, Kim KH, Kim YS, Woo J, Lee D, Son T, Kwon SW, Park KD, Yoon BE, Lee J, Li Y, Lee H, Bae YC, Lee CJ, Cheong E. Astrocytes Control Sensory Acuity via Tonic Inhibition in the Thalamus. Neuron 2020; 108:691-706.e10. [PMID: 32905785 DOI: 10.1016/j.neuron.2020.08.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/05/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022]
Abstract
Sensory discrimination is essential for survival. However, how sensory information is finely controlled in the brain is not well defined. Here, we show that astrocytes control tactile acuity via tonic inhibition in the thalamus. Mechanistically, diamine oxidase (DAO) and the subsequent aldehyde dehydrogenase 1a1 (Aldh1a1) convert putrescine into GABA, which is released via Best1. The GABA from astrocytes inhibits synaptically evoked firing at the lemniscal synapses to fine-tune the dynamic range of the stimulation-response relationship, the precision of spike timing, and tactile discrimination. Our findings reveal a novel role of astrocytes in the control of sensory acuity through tonic GABA release.
Collapse
Affiliation(s)
- Hankyul Kwak
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Wuhyun Koh
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea
| | - Sangwoo Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Kiyeong Song
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Jeong-Im Shin
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jung Moo Lee
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea
| | - Elliot H Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Jin Young Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41566, South Korea
| | - Go Eun Ha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Ju-Eun Oh
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yongmin Mason Park
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea
| | - Sunpil Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Ji Won Choi
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Ki Hun Kim
- Doping Control Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, College of Natural Science, Dankook University, Cheonan 31116, South Korea
| | - Junsung Woo
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Dongsu Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Taehwang Son
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, South Korea
| | - Soon Woo Kwon
- Radiation Medicine Clinical Research Division, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Ki Duk Park
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, South Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, South Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, College of Natural Science, Dankook University, Cheonan 31116, South Korea
| | - Jaeick Lee
- Doping Control Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Hyunbeom Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41566, South Korea
| | - C Justin Lee
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, South Korea; Center for Glia-Neuron Interaction, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea.
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea; POSTECH Biotech Center, POSTECH, Pohang, South Korea.
| |
Collapse
|
128
|
Live-imaging of astrocyte morphogenesis and function in zebrafish neural circuits. Nat Neurosci 2020; 23:1297-1306. [PMID: 32895565 PMCID: PMC7530038 DOI: 10.1038/s41593-020-0703-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/24/2020] [Indexed: 01/10/2023]
Abstract
How astrocytes grow and integrate into neural circuits remains poorly defined. Zebrafish are well-suited for such investigations, but bona fide astrocytes have not been described in this system. Here, we characterize a zebrafish cell type that is remarkably similar to mammalian astrocytes that derive from radial glial cells and elaborate processes to establish their territories at early larval stages. Zebrafish astrocytes associate closely with synapses, tile with one another, and express markers including Glast and glutamine synthetase. Once integrated into circuits, they exhibit whole-cell and microdomain Ca2+ transients, which are sensitive to norepinephrine. Finally, using a cell-specific CRISPR/Cas9 approach we demonstrate that fgfr3/4 are required for vertebrate astrocyte morphogenesis. This work provides the first visualization of astrocyte morphogenesis from stem cell to post-mitotic astrocyte in vivo, identifies a role for Fgf receptors in vertebrate astrocytes, and establishes zebrafish as a valuable new model system to study astrocyte biology in vivo.
Collapse
|
129
|
Verkhratsky A, Semyanov A, Zorec R. Physiology of Astroglial Excitability. FUNCTION (OXFORD, ENGLAND) 2020; 1:zqaa016. [PMID: 35330636 PMCID: PMC8788756 DOI: 10.1093/function/zqaa016] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023]
Abstract
Classic physiology divides all neural cells into excitable neurons and nonexcitable neuroglia. Neuroglial cells, chiefly responsible for homeostasis and defense of the nervous tissue, coordinate their complex homeostatic responses with neuronal activity. This coordination reflects a specific form of glial excitability mediated by complex changes in intracellular concentration of ions and second messengers organized in both space and time. Astrocytes are equipped with multiple molecular cascades, which are central for regulating homeostasis of neurotransmitters, ionostasis, synaptic connectivity, and metabolic support of the central nervous system. Astrocytes are further provisioned with multiple receptors for neurotransmitters and neurohormones, which upon activation trigger intracellular signals mediated by Ca2+, Na+, and cyclic AMP. Calcium signals have distinct organization and underlying mechanisms in different astrocytic compartments thus allowing complex spatiotemporal signaling. Signals mediated by fluctuations in cytosolic Na+ are instrumental for coordination of Na+ dependent astrocytic transporters with tissue state and homeostatic demands. Astroglial ionic excitability may also involve K+, H+, and Cl-. The cyclic AMP signalling system is, in comparison to ions, much slower in targeting astroglial effector mechanisms. This evidence review summarizes the concept of astroglial intracellular excitability.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK,Achucarro Center for Neuroscience, Ikerbasque, 48011 Bilbao, Spain,Address correspondence to A.V. (e-mail: )
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia,Faculty of Biology, Moscow State University, Moscow, Russia,Sechenov First Moscow State Medical University, Moscow, Russia
| | - Robert Zorec
- Celica Biomedical, Ljubljana 1000, Slovenia,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| |
Collapse
|
130
|
Semyanov A, Henneberger C, Agarwal A. Making sense of astrocytic calcium signals — from acquisition to interpretation. Nat Rev Neurosci 2020; 21:551-564. [DOI: 10.1038/s41583-020-0361-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
|
131
|
Deciphering the star codings: astrocyte manipulation alters mouse behavior. Exp Mol Med 2020; 52:1028-1038. [PMID: 32665584 PMCID: PMC8080576 DOI: 10.1038/s12276-020-0468-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 01/11/2023] Open
Abstract
Astrocytes occupy a vast area within the central nervous system (CNS). Despite their abundance, the functional role of astrocytes in vivo has only begun to be uncovered. Astrocytes were typically thought to be involved in pathophysiological states. However, recent studies have shown that astrocytes are actively involved in cell signaling in normal physiological states; manipulating various aspects of astrocytic cell signaling in vivo has revealed that astrocytes are key players in controlling healthy behavior in the absence of pathophysiology. Unfortunately, the study of astrocyte function is often limited by the number of approaches available due to our lack of understanding of cell physiology. This review summarizes recent studies in which altered astrocyte signaling capacity resulted in dramatic changes in behavior. We not only discuss the methodologies available to manipulate astrocytes but also provide insights into the behavioral roles of astrocytes in the CNS. Genetic studies provide increased evidence that astrocytes, star-shaped cells in the central nervous system, play important roles affecting behavior in mammals. Although they are just as abundant as neurons, astrocytes are not excited by electrical signals. For this reason they have traditionally been regarded simply as ‘support cells’ for neurons, but recent evidence suggests that they can significantly modulate neuron signals. A review paper by Keebum Park and Sung Joong Lee at Seoul National University in South Korea highlights improved methods for monitoring the signaling processes related to astrocytes, which manifest most notably through sharp changes in calcium levels. Several studies have used genetic knockout mice, designer drugs and light-sensitive proteins to change astrocyte activity, affecting a diverse range of behaviors including sleeping and feeding patterns, memory formation, depression and obsessive compulsive disorder.
Collapse
|
132
|
Bojarskaite L, Bjørnstad DM, Pettersen KH, Cunen C, Hermansen GH, Åbjørsbråten KS, Chambers AR, Sprengel R, Vervaeke K, Tang W, Enger R, Nagelhus EA. Astrocytic Ca 2+ signaling is reduced during sleep and is involved in the regulation of slow wave sleep. Nat Commun 2020; 11:3240. [PMID: 32632168 PMCID: PMC7338360 DOI: 10.1038/s41467-020-17062-2] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/09/2020] [Indexed: 12/31/2022] Open
Abstract
Astrocytic Ca2+ signaling has been intensively studied in health and disease but has not been quantified during natural sleep. Here, we employ an activity-based algorithm to assess astrocytic Ca2+ signals in the neocortex of awake and naturally sleeping mice while monitoring neuronal Ca2+ activity, brain rhythms and behavior. We show that astrocytic Ca2+ signals exhibit distinct features across the sleep-wake cycle and are reduced during sleep compared to wakefulness. Moreover, an increase in astrocytic Ca2+ signaling precedes transitions from slow wave sleep to wakefulness, with a peak upon awakening exceeding the levels during whisking and locomotion. Finally, genetic ablation of an important astrocytic Ca2+ signaling pathway impairs slow wave sleep and results in an increased number of microarousals, abnormal brain rhythms, and an increased frequency of slow wave sleep state transitions and sleep spindles. Our findings demonstrate an essential role for astrocytic Ca2+ signaling in regulating slow wave sleep.
Collapse
Affiliation(s)
- Laura Bojarskaite
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Daniel M Bjørnstad
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Klas H Pettersen
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Céline Cunen
- Statistics and Data Science group, Department of Mathematics, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316, Oslo, Norway
| | - Gudmund Horn Hermansen
- Statistics and Data Science group, Department of Mathematics, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316, Oslo, Norway
| | - Knut Sindre Åbjørsbråten
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Anna R Chambers
- Lab for Neural Computation, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Rolf Sprengel
- Research Group of the Max Planck Institute for Medical Research, Institute for Anatomy and Cell Biology, Heidelberg University, 69120, Heidelberg, Germany
| | - Koen Vervaeke
- Lab for Neural Computation, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Wannan Tang
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Enger
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway.
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway.
- Department of Neurology, Oslo University Hospital, Rikshospitalet, 0027, Oslo, Norway.
| | - Erlend A Nagelhus
- Letten Centre and GliaLab, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Rikshospitalet, 0027, Oslo, Norway
| |
Collapse
|
133
|
Samora M, Vianna LC, Carmo JC, Macedo V, Dawes M, Phillips AA, Paton JFR, Fisher JP. Neurovascular coupling is not influenced by lower body negative pressure in humans. Am J Physiol Heart Circ Physiol 2020; 319:H22-H31. [PMID: 32442032 DOI: 10.1152/ajpheart.00076.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cerebral blood flow is tightly coupled with local neuronal activation and metabolism, i.e., neurovascular coupling (NVC). Studies suggest a role of sympathetic nervous system in the regulation of cerebral blood flow. However, this is controversial, and the sympathetic regulation of NVC in humans remains unclear. Since impaired NVC has been identified in several chronic diseases associated with a heightened sympathetic activity, we aimed to determine whether reflex-mediated sympathetic activation via lower body negative pressure (LBNP) attenuates NVC in humans. NVC was assessed using a visual stimulation protocol (5 cycles of 30 s eyes closed and 30 s of reading) in 11 healthy participants (aged 24 ± 3 yr). NVC assessments were made under control conditions and during LBNP at -20 and -40 mmHg. Posterior (PCA) and middle (MCA) cerebral artery mean blood velocity (Vmean) and vertebral artery blood flow (VAflow) were simultaneously determined with cardiorespiratory variables. Under control conditions, the visual stimulation evoked a robust increase in PCAVmean (∆18.0 ± 4.5%), a moderate rise in VAflow (∆9.6 ± 4.3%), and a modest increase in MCAVmean (∆3.0 ± 1.9%). The magnitude of NVC response was not affected by mild-to-moderate LBNP (all P > 0.05 for repeated-measures ANOVA). Given the small change that occurred in partial pressure of end-tidal CO2 during LBNP, this hypocapnia condition was matched via voluntary hyperventilation in absence of LBNP in a subgroup of participants (n = 8). The mild hypocapnia during LBNP did not exert a confounding influence on the NVC response. These findings indicate that the NVC is not influenced by LBNP or mild hypocapnia in humans.NEW & NOTEWORTHY Visual stimulation evoked a robust increase in posterior cerebral artery velocity and a modest increase in vertebral artery blood flow, i.e., neurovascular coupling (NVC), which was unaffected by lower body negative pressure (LBNP) in humans. In addition, although LBNP induced a mild hypocapnia, this degree of hypocapnia in the absence of LBNP failed to modify the NVC response.
Collapse
Affiliation(s)
- Milena Samora
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,NeuroV̇ASQ̇-Integrative Physiology Laboratory, Faculty of Physical Education, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Lauro C Vianna
- NeuroV̇ASQ̇-Integrative Physiology Laboratory, Faculty of Physical Education, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Jake C Carmo
- Biomechanics and Biological Signal Processing Laboratory, Faculty of Physical Education, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Victor Macedo
- Biomechanics and Biological Signal Processing Laboratory, Faculty of Physical Education, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Matthew Dawes
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Aaron A Phillips
- Departments of Physiology, Pharmacology, and Clinical Neurosciences, Libin Cardiovascular Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Julian F R Paton
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - James P Fisher
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
134
|
Pham C, Moro DH, Mouffle C, Didienne S, Hepp R, Pfrieger FW, Mangin JM, Legendre P, Martin C, Luquet S, Cauli B, Li D. Mapping astrocyte activity domains by light sheet imaging and spatio-temporal correlation screening. Neuroimage 2020; 220:117069. [PMID: 32585347 DOI: 10.1016/j.neuroimage.2020.117069] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
Astrocytes are a major type of glial cell in the mammalian brain, essentially regulating neuronal development and function. Quantitative imaging represents an important approach to study astrocytic signaling in neural circuits. Focusing on astrocytic Ca2+ activity, a key pathway implicated in astrocye-neuron interaction, we here report a strategy combining fast light sheet fluorescence microscopy (LSFM) and correlative screening-based time series analysis, to map activity domains in astrocytes in living mammalian nerve tissue. Light sheet of micron-scale thickness enables wide-field optical sectioning to image astrocytes in acute mouse brain slices. Using both chemical and genetically encoded Ca2+ indicators, we demonstrate the complementary advantages of LSFM in mapping Ca2+ domains in astrocyte populations as compared to epifluorescence and two-photon microscopy. Our approach then revealed distinct kinetics of Ca2+ signals between cortical and hypothalamic astrocytes in resting conditions and following the activation of adrenergic G protein coupled receptor (GPCR). This observation highlights the activity heterogeneity across regionally distinct astrocyte populations, and indicates the potential of our method for investigating dynamic signals in astrocytes.
Collapse
Affiliation(s)
- Cuong Pham
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Daniela Herrera Moro
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, Unité Mixte de Recherche 8251, Université Paris Diderot, Sorbonne Paris Cité, 75205, Paris, France
| | - Christine Mouffle
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Steve Didienne
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Régine Hepp
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Frank W Pfrieger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000, Strasbourg, France
| | - Jean-Marie Mangin
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Pascal Legendre
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Claire Martin
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, Unité Mixte de Recherche 8251, Université Paris Diderot, Sorbonne Paris Cité, 75205, Paris, France
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, Unité Mixte de Recherche 8251, Université Paris Diderot, Sorbonne Paris Cité, 75205, Paris, France
| | - Bruno Cauli
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Dongdong Li
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France.
| |
Collapse
|
135
|
Gaidin SG, Zinchenko VP, Sergeev AI, Teplov IY, Mal'tseva VN, Kosenkov AM. Activation of alpha‐2 adrenergic receptors stimulates GABA release by astrocytes. Glia 2020; 68:1114-1130. [DOI: 10.1002/glia.23763] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Sergei G. Gaidin
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences” Pushchino Russia
| | - Valery P. Zinchenko
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences” Pushchino Russia
| | - Alexander I. Sergeev
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences” Pushchino Russia
| | - Ilia Y. Teplov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences” Pushchino Russia
| | - Valentina N. Mal'tseva
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences” Pushchino Russia
| | - Artem M. Kosenkov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences” Pushchino Russia
| |
Collapse
|
136
|
Abstract
Astrocytes are morphologically complex, ubiquitous cells that are viewed as a homogeneous population tiling the entire central nervous system (CNS). However, this view has been challenged in the last few years with the availability of RNA sequencing, immunohistochemistry, electron microscopy, morphological reconstruction, and imaging data. These studies suggest that astrocytes represent a diverse population of cells and that they display brain area- and disease-specific properties and functions. In this review, we summarize these observations, emphasize areas where clear conclusions can be made, and discuss potential unifying themes. We also identify knowledge gaps that need to be addressed in order to exploit astrocyte diversity as a biological phenomenon of physiological relevance in the CNS. We thus provide a summary and a perspective on astrocyte diversity in the vertebrate CNS.
Collapse
Affiliation(s)
- Baljit S Khakh
- Departments of Physiology and Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA;
| | - Benjamin Deneen
- Department of Neuroscience and Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
137
|
Velebit J, Horvat A, Smolič T, Prpar Mihevc S, Rogelj B, Zorec R, Vardjan N. Astrocytes with TDP-43 inclusions exhibit reduced noradrenergic cAMP and Ca 2+ signaling and dysregulated cell metabolism. Sci Rep 2020; 10:6003. [PMID: 32265469 PMCID: PMC7138839 DOI: 10.1038/s41598-020-62864-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Most cases of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have cytoplasmic inclusions of TAR DNA-binding protein 43 (TDP-43) in neurons and non-neuronal cells, including astrocytes, which metabolically support neurons with nutrients. Neuronal metabolism largely depends on the activation of the noradrenergic system releasing noradrenaline. Activation of astroglial adrenergic receptors with noradrenaline triggers cAMP and Ca2+ signaling and augments aerobic glycolysis with production of lactate, an important neuronal energy fuel. Astrocytes with cytoplasmic TDP-43 inclusions can cause motor neuron death, however, whether astroglial metabolism and metabolic support of neurons is altered in astrocytes with TDP-43 inclusions, is unclear. We measured lipid droplet and glucose metabolisms in astrocytes expressing the inclusion-forming C-terminal fragment of TDP-43 or the wild-type TDP-43 using fluorescent dyes or genetically encoded nanosensors. Astrocytes with TDP-43 inclusions exhibited a 3-fold increase in the accumulation of lipid droplets versus astrocytes expressing wild-type TDP-43, indicating altered lipid droplet metabolism. In these cells the noradrenaline-triggered increases in intracellular cAMP and Ca2+ levels were reduced by 35% and 31%, respectively, likely due to the downregulation of β2-adrenergic receptors. Although noradrenaline triggered a similar increase in intracellular lactate levels in astrocytes with and without TDP-43 inclusions, the probability of activating aerobic glycolysis was facilitated by 1.6-fold in astrocytes with TDP-43 inclusions and lactate MCT1 transporters were downregulated. Thus, while in astrocytes with TDP-43 inclusions noradrenergic signaling is reduced, aerobic glycolysis and lipid droplet accumulation are facilitated, suggesting dysregulated astroglial metabolism and metabolic support of neurons in TDP-43-associated ALS and FTD.
Collapse
Affiliation(s)
- Jelena Velebit
- Laboratory of Cell Engineering, Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Anemari Horvat
- Laboratory of Cell Engineering, Celica Biomedical, 1000, Ljubljana, Slovenia.,Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Tina Smolič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Sonja Prpar Mihevc
- Department of Biotechnology, Jožef Stefan Institute, 1000, Ljubljana, Slovenia
| | - Boris Rogelj
- Department of Biotechnology, Jožef Stefan Institute, 1000, Ljubljana, Slovenia.,Biomedical Research Institute BRIS, 1000, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000, Ljubljana, Slovenia.,Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, 1000, Ljubljana, Slovenia. .,Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia.
| |
Collapse
|
138
|
Emotional Stress Induces Structural Plasticity in Bergmann Glial Cells via an AC5-CPEB3-GluA1 Pathway. J Neurosci 2020; 40:3374-3384. [PMID: 32229518 DOI: 10.1523/jneurosci.0013-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/13/2020] [Accepted: 02/27/2020] [Indexed: 11/21/2022] Open
Abstract
Stress alters brain function by modifying the structure and function of neurons and astrocytes. The fine processes of astrocytes are critical for the clearance of neurotransmitters during synaptic transmission. Thus, experience-dependent remodeling of glial processes is anticipated to alter the output of neural circuits. However, the molecular mechanisms that underlie glial structural plasticity are not known. Here we show that a single exposure of male and female mice to an acute stress produced a long-lasting retraction of the lateral processes of cerebellar Bergmann glial cells. These cells express the GluA1 subunit of AMPA-type glutamate receptors, and GluA1 knockdown is known to shorten the length of glial processes. We found that stress reduced the level of GluA1 protein and AMPA receptor-mediated currents in Bergmann glial cells, and these effects were absent in mice devoid of CPEB3, a protein that binds to GluA1 mRNA and regulates GluA1 protein synthesis. Administration of a β-adrenergic receptor blocker attenuated the reduction in GluA1, and deletion of adenylate cyclase 5 prevented GluA1 suppression. Therefore, stress suppresses GluA1 protein synthesis via an adrenergic/adenylyl cyclase/CPEB3 pathway, and reduces the length of astrocyte lateral processes. Our results identify a novel mechanism for GluA1 subunit plasticity in non-neuronal cells and suggest a previously unappreciated role for AMPA receptors in stress-induced astrocytic remodeling.SIGNIFICANCE STATEMENT Astrocytes play important roles in synaptic transmission by extending fine processes around synapses. In this study, we showed that a single exposure to an acute stress triggered a retraction of lateral/fine processes in mouse cerebellar astrocytes. These astrocytes express GluA1, a glutamate receptor subunit known to lengthen astrocyte processes. We showed that astrocytic structural changes are associated with a reduction of GluA1 protein levels. This requires activation of β-adrenergic receptors and is triggered by noradrenaline released during stress. We identified adenylyl cyclase 5, an enzyme that elevates cAMP levels, as a downstream effector and found that lowering GluA1 levels depends on CPEB3 proteins that bind to GluA1 mRNA. Therefore, stress regulates GluA1 protein synthesis via an adrenergic/adenylyl cyclase/CPEB3 pathway in astrocytes and remodels their fine processes.
Collapse
|
139
|
Kofuji P, Araque A. G-Protein-Coupled Receptors in Astrocyte-Neuron Communication. Neuroscience 2020; 456:71-84. [PMID: 32224231 DOI: 10.1016/j.neuroscience.2020.03.025] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022]
Abstract
Astrocytes, a major type of glial cell, are known to play key supportive roles in brain function, contributing to ion and neurotransmitter homeostasis, maintaining the blood-brain barrier and providing trophic and metabolic support for neurons. Besides these support functions, astrocytes are emerging as important elements in brain physiology through signaling exchange with neurons at tripartite synapses. Astrocytes express a wide variety of neurotransmitter transporters and receptors that allow them to sense and respond to synaptic activity. Principal among them are the G-protein-coupled receptors (GPCRs) in astrocytes because their activation by synaptically released neurotransmitters leads to mobilization of intracellular calcium. In turn, activated astrocytes release neuroactive substances called gliotransmitters, such as glutamate, GABA, and ATP/adenosine that lead to synaptic regulation through activation of neuronal GPCRs. In this review we will present and discuss recent evidence demonstrating the critical roles played by GPCRs in the bidirectional astrocyte-neuron signaling, and their crucial involvement in the astrocyte-mediated regulation of synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
140
|
Pais-Roldán P, Takahashi K, Sobczak F, Chen Y, Zhao X, Zeng H, Jiang Y, Yu X. Indexing brain state-dependent pupil dynamics with simultaneous fMRI and optical fiber calcium recording. Proc Natl Acad Sci U S A 2020; 117:6875-6882. [PMID: 32139609 PMCID: PMC7104268 DOI: 10.1073/pnas.1909937117] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pupillometry, a noninvasive measure of arousal, complements human functional MRI (fMRI) to detect periods of variable cognitive processing and identify networks that relate to particular attentional states. Even under anesthesia, pupil dynamics correlate with brain-state fluctuations, and extended dilations mark the transition to more arousable states. However, cross-scale neuronal activation patterns are seldom linked to brain state-dependent pupil dynamics. Here, we complemented resting-state fMRI in rats with cortical calcium recording (GCaMP-mediated) and pupillometry to tackle the linkage between brain-state changes and neural dynamics across different scales. This multimodal platform allowed us to identify a global brain network that covaried with pupil size, which served to generate an index indicative of the brain-state fluctuation during anesthesia. Besides, a specific correlation pattern was detected in the brainstem, at a location consistent with noradrenergic cell group 5 (A5), which appeared to be dependent on the coupling between different frequencies of cortical activity, possibly further indicating particular brain-state dynamics. The multimodal fMRI combining concurrent calcium recordings and pupillometry enables tracking brain state-dependent pupil dynamics and identifying unique cross-scale neuronal dynamic patterns under anesthesia.
Collapse
Affiliation(s)
- Patricia Pais-Roldán
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tuebingen, 72074 Tuebingen, Germany
- Medical Imaging Physics, Institute of Neuroscience and Medicine, Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Kengo Takahashi
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tuebingen, 72074 Tuebingen, Germany
| | - Filip Sobczak
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tuebingen, 72074 Tuebingen, Germany
| | - Yi Chen
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tuebingen, 72074 Tuebingen, Germany
| | - Xiaoning Zhao
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
| | - Hang Zeng
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tuebingen, 72074 Tuebingen, Germany
| | - Yuanyuan Jiang
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Xin Yu
- High-Field Magnetic Resonance Department, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany;
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| |
Collapse
|
141
|
King CM, Bohmbach K, Minge D, Delekate A, Zheng K, Reynolds J, Rakers C, Zeug A, Petzold GC, Rusakov DA, Henneberger C. Local Resting Ca 2+ Controls the Scale of Astroglial Ca 2+ Signals. Cell Rep 2020; 30:3466-3477.e4. [PMID: 32160550 PMCID: PMC7068654 DOI: 10.1016/j.celrep.2020.02.043] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/21/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Astroglia regulate neurovascular coupling while engaging in signal exchange with neurons. The underlying cellular machinery is thought to rely on astrocytic Ca2+ signals, but what controls their amplitude and waveform is poorly understood. Here, we employ time-resolved two-photon excitation fluorescence imaging in acute hippocampal slices and in cortex in vivo to find that resting [Ca2+] predicts the scale (amplitude) and the maximum (peak) of astroglial Ca2+ elevations. We bidirectionally manipulate resting [Ca2+] by uncaging intracellular Ca2+ or Ca2+ buffers and use ratiometric imaging of a genetically encoded Ca2+ indicator to establish that alterations in resting [Ca2+] change co-directionally the peak level and anti-directionally the amplitude of local Ca2+ transients. This relationship holds for spontaneous and for induced (for instance by locomotion) Ca2+ signals. Our findings uncover a basic generic rule of Ca2+ signal formation in astrocytes, thus also associating the resting Ca2+ level with the physiological "excitability" state of astroglia.
Collapse
Affiliation(s)
- Claire M King
- Institute of Neurology, University College London, London, UK
| | - Kirsten Bohmbach
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Daniel Minge
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andrea Delekate
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kaiyu Zheng
- Institute of Neurology, University College London, London, UK
| | - James Reynolds
- Institute of Neurology, University College London, London, UK
| | - Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurology, University Hospital Bonn, Bonn, Germany
| | | | - Christian Henneberger
- Institute of Neurology, University College London, London, UK; Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
142
|
Wotton CA, Cross CD, Bekar LK. Serotonin, norepinephrine, and acetylcholine differentially affect astrocytic potassium clearance to modulate somatosensory signaling in male mice. J Neurosci Res 2020; 98:964-977. [PMID: 32067254 DOI: 10.1002/jnr.24597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/21/2020] [Accepted: 02/01/2020] [Indexed: 11/07/2022]
Abstract
Changes in extracellular potassium ([K+ ]e ) modulate neuronal networks via changes in membrane potential, voltage-gated channel activity, and alteration to transmission at the synapse. Given the limited extracellular space in the central nervous system, potassium clearance is crucial. As activity-induced potassium transients are rapidly managed by astrocytic Kir4.1 and astrocyte-specific Na+ /K+ -ATPase, any neurotransmitter/neuromodulator that can regulate their function may have indirect influence on network activity. Neuromodulators differentially affect cortical/thalamic networks to align sensory processing with differing behavioral states. Given serotonin (5HT), norepinephrine (NE), and acetylcholine (ACh) differentially affect spike frequency adaptation and signal fidelity ("signal-to-noise") in somatosensory cortex, we hypothesize that [K+ ]e may be differentially regulated by the different neuromodulators to exert their individual effects on network function. This study aimed to compare effects of individually applied 5HT, NE, and ACh on regulating [K+ ]e in connection to effects on cortical-evoked response amplitude and adaptation in male mice. Using extracellular field and K+ ion-selective recordings of somatosensory stimulation, we found that differential effects of 5HT, NE, and ACh on [K+ ]e regulation mirrored differential effects on amplitude and adaptation. 5HT effects on transient K+ recovery, adaptation, and field post-synaptic potential amplitude were disrupted by barium (200 µM), whereas NE and ACh effects were disrupted by ouabain (1 µM) or iodoacetate (100 µM). Considering the impact [K+ ]e can have on many network functions; it seems highly efficient that neuromodulators regulate [K+ ]e to exert their many effects. This study provides functional significance for astrocyte-mediated buffering of [K+ ]e in neuromodulator-mediated shaping of cortical network activity.
Collapse
Affiliation(s)
- Caitlin A Wotton
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Cassidy D Cross
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lane K Bekar
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
143
|
Yu X, Nagai J, Khakh BS. Improved tools to study astrocytes. Nat Rev Neurosci 2020; 21:121-138. [DOI: 10.1038/s41583-020-0264-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
144
|
Zuend M, Saab AS, Wyss MT, Ferrari KD, Hösli L, Looser ZJ, Stobart JL, Duran J, Guinovart JJ, Barros LF, Weber B. Arousal-induced cortical activity triggers lactate release from astrocytes. Nat Metab 2020; 2:179-191. [PMID: 32694692 DOI: 10.1038/s42255-020-0170-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 01/15/2020] [Indexed: 01/01/2023]
Abstract
It has been suggested that, in states of arousal, release of noradrenaline and β-adrenergic signalling affect long-term memory formation by stimulating astrocytic lactate production from glycogen. However, the temporal relationship between cortical activity and cellular lactate fluctuations upon changes in arousal remains to be fully established. Also, the role of β-adrenergic signalling and brain glycogen metabolism on neural lactate dynamics in vivo is still unknown. Here, we show that an arousal-induced increase in cortical activity triggers lactate release into the extracellular space, and this correlates with a fast and prominent lactate dip in astrocytes. The immediate drop in astrocytic lactate concentration and the parallel increase in extracellular lactate levels underline an activity-dependent lactate release from astrocytes. Moreover, when β-adrenergic signalling is blocked or the brain is depleted of glycogen, the arousal-evoked cellular lactate surges are significantly reduced. We provide in vivo evidence that cortical activation upon arousal triggers lactate release from astrocytes, a rise in intracellular lactate levels mediated by β-adrenergic signalling and the mobilization of lactate from glycogen stores.
Collapse
Affiliation(s)
- Marc Zuend
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Matthias T Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ladina Hösli
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jordi Duran
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
| | | | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
145
|
Malan L, Hamer M, von Känel R, van Wyk RD, Wentzel A, Steyn HS, van Vuuren P, Malan NT. Retinal-glia ischemia and inflammation induced by chronic stress: The SABPA study. Brain Behav Immun Health 2020; 2:100027. [PMID: 38377420 PMCID: PMC8474432 DOI: 10.1016/j.bbih.2019.100027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/14/2019] [Accepted: 12/15/2019] [Indexed: 01/10/2023] Open
Abstract
Background Psychobiological processes linking stress and vascular diseases remain poorly understood. The retina and the brain share a common embryonic-diencephalon origin and blood-barrier physiology e.g. ongoing ischemia facilitates S100B release with astrocytic activity and glial-fibrillary-acidic-protein expression (GFAP). However, GFAP decreases revealed astrocyte pathology in the prefrontal cortex of depression/suicide cases; and might be a key mechanism in stress - disease pathways. Methods A chronic emotional stress phenotype independent of age, ethnicity or sex was used to stratify the current prospective cohort (N = 359; aged 46 ± 9 years) into Stress (N = 236) and no-Stress groups (N = 123). Prospective data for glia ischemia risk markers were obtained, including 24 h BP, fasting S100B, GFAP, HbA1C and tumor-necrosis-factor-α (TNF-α). At 3-yr follow-up: diastolic-ocular-perfusion-pressure (indicating hypo-perfusion risk) was measured and retinal vessel calibers were quantified from digital images in the mydriatic eye. Results Higher hypertension (75% vs. 16%), diabetes (13% vs. 0%) and retinopathy (57% vs. 45%) prevalence was observed in Stress compared to no-Stress individuals. Stressed individuals had consistently raised S100B, TNF-α, HbA1C and higher diastolic-ocular-perfusion-pressure, but decreases in GFAP and GFAP:S100B. Furthermore stroke risk markers, arterial narrowing and venous widening were associated with consistently raised S100B, GFAP:S100B (p = 0.060), TNF-α and higher diastolic-ocular-perfusion-pressure [Adj. R2 0.39-0.41, p ≤ 0.05]. No retinal-glia associations were evident in the no-Stress group. Conclusions Retinal-glia ischemia and inflammation was induced by chronic stress. Persistent higher inflammation and S100B with GFAP decreases further reflected stress-induced astrocyte pathology in the human retina. It is recommended to increase awareness on chronic stress and susceptibility for brain ischemia.
Collapse
Affiliation(s)
- Leoné Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, 2520, South Africa
| | - Mark Hamer
- Division Surgery & Interventional Science, University College London, United Kingdom
| | - Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, 8091, Switzerland
| | - Roelof D. van Wyk
- Surgical Ophthalmologist, 85 Peter Mokaba Street, Potchefstroom, 2531, South Africa
| | - Annemarie Wentzel
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, 2520, South Africa
| | - Hendrik S. Steyn
- Statistical Consultation Services, North-West University, Potchefstroom, 2520, South Africa
| | - Pieter van Vuuren
- School of Electrical, Electronic and Computer Engineering, North-West University, Potchefstroom, 2520, South Africa
| | - Nico T. Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, 2520, South Africa
| |
Collapse
|
146
|
Distinct temporal integration of noradrenaline signaling by astrocytic second messengers during vigilance. Nat Commun 2020; 11:471. [PMID: 31980655 PMCID: PMC6981284 DOI: 10.1038/s41467-020-14378-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/17/2019] [Indexed: 01/12/2023] Open
Abstract
Astrocytes may function as mediators of the impact of noradrenaline on neuronal function. Activation of glial α1-adrenergic receptors triggers rapid astrocytic Ca2+ elevation and facilitates synaptic plasticity, while activation of β-adrenergic receptors elevates cAMP levels and modulates memory consolidation. However, the dynamics of these processes in behaving mice remain unexplored, as do the interactions between the distinct second messenger pathways. Here we simultaneously monitored astrocytic Ca2+ and cAMP and demonstrate that astrocytic second messengers are regulated in a temporally distinct manner. In behaving mice, we found that while an abrupt facial air puff triggered transient increases in noradrenaline release and large cytosolic astrocytic Ca2+ elevations, cAMP changes were not detectable. By contrast, repeated aversive stimuli that lead to prolonged periods of vigilance were accompanied by robust noradrenergic axonal activity and gradual sustained cAMP increases. Our findings suggest distinct astrocytic signaling pathways can integrate noradrenergic activity during vigilance states to mediate distinct functions supporting memory. Astrocytic GPCRs activate Ca2+ and cAMP signaling pathways, however, the in vivo dynamics of the two second messengers have not been fully been characterized. The authors demonstrate distinct noradrenaline-induced astrocytic Ca2+ and cAMP dynamics during startle and fear conditioning.
Collapse
|
147
|
Kastanenka KV, Moreno-Bote R, De Pittà M, Perea G, Eraso-Pichot A, Masgrau R, Poskanzer KE, Galea E. A roadmap to integrate astrocytes into Systems Neuroscience. Glia 2020; 68:5-26. [PMID: 31058383 PMCID: PMC6832773 DOI: 10.1002/glia.23632] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
Systems neuroscience is still mainly a neuronal field, despite the plethora of evidence supporting the fact that astrocytes modulate local neural circuits, networks, and complex behaviors. In this article, we sought to identify which types of studies are necessary to establish whether astrocytes, beyond their well-documented homeostatic and metabolic functions, perform computations implementing mathematical algorithms that sub-serve coding and higher-brain functions. First, we reviewed Systems-like studies that include astrocytes in order to identify computational operations that these cells may perform, using Ca2+ transients as their encoding language. The analysis suggests that astrocytes may carry out canonical computations in a time scale of subseconds to seconds in sensory processing, neuromodulation, brain state, memory formation, fear, and complex homeostatic reflexes. Next, we propose a list of actions to gain insight into the outstanding question of which variables are encoded by such computations. The application of statistical analyses based on machine learning, such as dimensionality reduction and decoding in the context of complex behaviors, combined with connectomics of astrocyte-neuronal circuits, is, in our view, fundamental undertakings. We also discuss technical and analytical approaches to study neuronal and astrocytic populations simultaneously, and the inclusion of astrocytes in advanced modeling of neural circuits, as well as in theories currently under exploration such as predictive coding and energy-efficient coding. Clarifying the relationship between astrocytic Ca2+ and brain coding may represent a leap forward toward novel approaches in the study of astrocytes in health and disease.
Collapse
Affiliation(s)
- Ksenia V. Kastanenka
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Massachusetts 02129, USA
| | - Rubén Moreno-Bote
- Department of Information and Communications Technologies, Center for Brain and Cognition and Universitat Pompeu Fabra, 08018 Barcelona, Spain
- ICREA, 08010 Barcelona, Spain
| | | | | | - Abel Eraso-Pichot
- Departament de Bioquímica, Institut de Neurociències i Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Roser Masgrau
- Departament de Bioquímica, Institut de Neurociències i Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Kira E. Poskanzer
- Department of Biochemistry & Biophysics, Neuroscience Graduate Program, and Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California 94143, USA
- Equally contributing authors
| | - Elena Galea
- ICREA, 08010 Barcelona, Spain
- Departament de Bioquímica, Institut de Neurociències i Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Equally contributing authors
| |
Collapse
|
148
|
Okubo Y, Iino M, Hirose K. Store-operated Ca 2+ entry-dependent Ca 2+ refilling in the endoplasmic reticulum in astrocytes. Biochem Biophys Res Commun 2019; 522:1003-1008. [PMID: 31812243 DOI: 10.1016/j.bbrc.2019.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022]
Abstract
Astrocytes regulate various brain functions, for which Ca2+ release from the endoplasmic reticulum (ER) often play crucial roles. Because astrocytic ER Ca2+ release is robust and frequent, the ER Ca2+ refilling mechanism should be critical for ongoing Ca2+ signaling in astrocytes. In this study, we focused on the putative functional significance of store-operated Ca2+ entry (SOCE) in ER Ca2+ refilling. We expressed the ER luminal Ca2+ indicator G-CEPIA1er in astrocytes in acute cortical slices to directly monitor the decrease and recovery of ER Ca2+ concentration upon spontaneous or norepinephrine-induced Ca2+ release. Inhibition of SOCE significantly slowed the recovery of ER Ca2+ concentration after Ca2+ release in astrocytes. This delayed recovery resulted in a prolonged decrease in the ER Ca2+ content in astrocytes with periodic spontaneous Ca2+ release, followed by the attenuation of cytosolic Ca2+ responses upon Ca2+ release. Therefore, our results provide direct evidence for the physiological significance of SOCE in ER Ca2+ refilling after ER Ca2+ release.
Collapse
Affiliation(s)
- Yohei Okubo
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 133-0033, Japan.
| | - Masamitsu Iino
- Division of Cellular and Molecular Pharmacology, Nihon University School of Medicine, 30-1 Oyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 133-0033, Japan
| |
Collapse
|
149
|
Stowell RD, Sipe GO, Dawes RP, Batchelor HN, Lordy KA, Whitelaw BS, Stoessel MB, Bidlack JM, Brown E, Sur M, Majewska AK. Noradrenergic signaling in the wakeful state inhibits microglial surveillance and synaptic plasticity in the mouse visual cortex. Nat Neurosci 2019; 22:1782-1792. [PMID: 31636451 PMCID: PMC6875777 DOI: 10.1038/s41593-019-0514-0] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/12/2019] [Indexed: 12/28/2022]
Abstract
Microglia are the brain's resident innate immune cells and also have a role in synaptic plasticity. Microglial processes continuously survey the brain parenchyma, interact with synaptic elements and maintain tissue homeostasis. However, the mechanisms that control surveillance and its role in synaptic plasticity are poorly understood. Microglial dynamics in vivo have been primarily studied in anesthetized animals. Here we report that microglial surveillance and injury response are reduced in awake mice as compared to anesthetized mice, suggesting that arousal state modulates microglial function. Pharmacologic stimulation of β2-adrenergic receptors recapitulated these observations and disrupted experience-dependent plasticity, and these effects required the presence of β2-adrenergic receptors in microglia. These results indicate that microglial roles in surveillance and synaptic plasticity in the mouse brain are modulated by noradrenergic tone fluctuations between arousal states and emphasize the need to understand the effect of disruptions of adrenergic signaling in neurodevelopment and neuropathology.
Collapse
Affiliation(s)
- Rianne D Stowell
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Neuroscience Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Grayson O Sipe
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan P Dawes
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Neuroscience Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Hanna N Batchelor
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Katheryn A Lordy
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Brendan S Whitelaw
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Neuroscience Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Mark B Stoessel
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Neuroscience Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Jean M Bidlack
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward Brown
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
150
|
Buskila Y, Bellot-Saez A, Morley JW. Generating Brain Waves, the Power of Astrocytes. Front Neurosci 2019; 13:1125. [PMID: 31680846 PMCID: PMC6813784 DOI: 10.3389/fnins.2019.01125] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022] Open
Abstract
Synchronization of neuronal activity in the brain underlies the emergence of neuronal oscillations termed “brain waves”, which serve various physiological functions and correlate with different behavioral states. It has been postulated that at least ten distinct mechanisms are involved in the formulation of these brain waves, including variations in the concentration of extracellular neurotransmitters and ions, as well as changes in cellular excitability. In this mini review we highlight the contribution of astrocytes, a subtype of glia, in the formation and modulation of brain waves mainly due to their close association with synapses that allows their bidirectional interaction with neurons, and their syncytium-like activity via gap junctions that facilitate communication to distal brain regions through Ca2+ waves. These capabilities allow astrocytes to regulate neuronal excitability via glutamate uptake, gliotransmission and tight control of the extracellular K+ levels via a process termed K+ clearance. Spatio-temporal synchrony of activity across neuronal and astrocytic networks, both locally and distributed across cortical regions, underpins brain states and thereby behavioral states, and it is becoming apparent that astrocytes play an important role in the development and maintenance of neural activity underlying these complex behavioral states.
Collapse
Affiliation(s)
- Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW, Australia
| | - Alba Bellot-Saez
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW, Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|